51
|
Sánchez-Romero N, Schophuizen CM, Giménez I, Masereeuw R. In vitro systems to study nephropharmacology: 2D versus 3D models. Eur J Pharmacol 2016; 790:36-45. [DOI: 10.1016/j.ejphar.2016.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/24/2016] [Accepted: 07/06/2016] [Indexed: 12/20/2022]
|
52
|
Chiang IN, Huang WC, Huang CY, Pu YS, Young TH. Development of a chitosan-based tissue-engineered renal proximal tubule conduit. J Biomed Mater Res B Appl Biomater 2016; 106:9-20. [PMID: 27801972 DOI: 10.1002/jbm.b.33808] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 09/21/2016] [Accepted: 10/12/2016] [Indexed: 11/10/2022]
Abstract
Renal proximal tubule cells (RPTCs) are responsible for glomerular filtration and maintenance of water/electrolyte balance. To regenerate a proximal tubule, sufficient cell numbers and normal cell function are requisite. Collagen has been routinely used as a substrate for culturing human RPTCs (HRPTCs); however, the role of biomaterials has not been thoroughly explored. In this study, RPTCs retrieved from human nephrectomy/nephroureterectomy specimens were cultivated on chitosan as a substrate in serum-free condition for up to 150 days. HRPTCs could maintain a typical epithelial morphology and the specific differentiation feature of transporting epithelia after such long-term culture. As compared with HRPTCs cultivated on collagen, those cultivated on chitosan showed more dome formation, higher Na+ -K+ ATPase expression, lower vimentin expression, and lower transepithelial electrical resistance, indicating that HRPTCs cultivated on chitosan presented better differentiation status and would be more functional with better active transportation. Thus, the current study indicates greater scope for the use of chitosan as a biomaterial for preparing a HRPTC-coated chitosan conduit, which might be useful for the scaffold design of tissue-engineered proximal tubules. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 9-20, 2018.
Collapse
Affiliation(s)
- I-Ni Chiang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan.,Department of Urology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Chen Huang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Chao-Yuan Huang
- Department of Urology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yeong-Shiau Pu
- Department of Urology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tai-Horng Young
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
53
|
Abstract
Treatment and management of kidney disease currently presents an enormous global burden, and the application of nanotechnology principles to renal disease therapy, although still at an early stage, has profound transformative potential. The increasing translation of nanomedicines to the clinic, alongside research efforts in tissue regeneration and organ-on-a-chip investigations, are likely to provide novel solutions to treat kidney diseases. Our understanding of renal anatomy and of how the biological and physico-chemical properties of nanomedicines (the combination of a nanocarrier and a drug) influence their interactions with renal tissues has improved dramatically. Tailoring of nanomedicines in terms of kidney retention and binding to key membranes and cell populations associated with renal diseases is now possible and greatly enhances their localization, tolerability, and efficacy. This Review outlines nanomedicine characteristics central to improved targeting of renal cells and highlights the prospects, challenges, and opportunities of nanotechnology-mediated therapies for renal diseases.
Collapse
|
54
|
Du C, Narayanan K, Leong MF, Ibrahim MS, Chua YP, Khoo VMH, Wan ACA. Functional Kidney Bioengineering with Pluripotent Stem-Cell-Derived Renal Progenitor Cells and Decellularized Kidney Scaffolds. Adv Healthc Mater 2016; 5:2080-91. [PMID: 27294565 DOI: 10.1002/adhm.201600120] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/20/2016] [Indexed: 11/11/2022]
Abstract
Recent advances in developmental biology and stem cell technology have led to the engineering of functional organs in a dish. However, the limited size of these organoids and absence of a large circulatory system poses limits to its clinical translation. To overcome these issues, decellularized whole kidney scaffolds with native microstructure and extracellular matrix (ECM) are employed for kidney bioengineering, using human-induced pluripotent-stem-cell-derived renal progenitor cells and endothelial cells. To demonstrate ECM-guided cellular assembly, the present work is focused on generating the functional unit of the kidney, the glomerulus. In the repopulated organ, the presence of endothelial cells broadly upregulates the expression level of genes related to renal development. When the cellularized native scaffolds are implanted in SCID mice, glomeruli assembly can be achieved by co-culture of the renal progenitors and endothelial cells. These individual glomerular units are shown to be functional in the context of the whole organ using a simulated bio-reactor set-up with urea and creatinine excretion and albumin reabsorption. Our results indicate that the repopulation of decellularized native kidney using clinically relevant, expandable patient-specific renal progenitors and endothelial cells may be a viable approach for the generation of a functional whole kidney.
Collapse
Affiliation(s)
- Chan Du
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | - Karthikeyan Narayanan
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | - Meng Fatt Leong
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | | | - Ying Ping Chua
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | - Vanessa Mei Hui Khoo
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | - Andrew C. A. Wan
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| |
Collapse
|
55
|
Paoli R, Samitier J. Mimicking the Kidney: A Key Role in Organ-on-Chip Development. MICROMACHINES 2016; 7:E126. [PMID: 30404298 PMCID: PMC6190229 DOI: 10.3390/mi7070126] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 12/29/2022]
Abstract
Pharmaceutical drug screening and research into diseases call for significant improvement in the effectiveness of current in vitro models. Better models would reduce the likelihood of costly failures at later drug development stages, while limiting or possibly even avoiding the use of animal models. In this regard, promising advances have recently been made by the so-called "organ-on-chip" (OOC) technology. By combining cell culture with microfluidics, biomedical researchers have started to develop microengineered models of the functional units of human organs. With the capacity to mimic physiological microenvironments and vascular perfusion, OOC devices allow the reproduction of tissue- and organ-level functions. When considering drug testing, nephrotoxicity is a major cause of attrition during pre-clinical, clinical, and post-approval stages. Renal toxicity accounts for 19% of total dropouts during phase III drug evaluation-more than half the drugs abandoned because of safety concerns. Mimicking the functional unit of the kidney, namely the nephron, is therefore a crucial objective. Here we provide an extensive review of the studies focused on the development of a nephron-on-chip device.
Collapse
Affiliation(s)
- Roberto Paoli
- Nanobioengineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), Barcelona 08028, Spain.
| | - Josep Samitier
- Nanobioengineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), Barcelona 08028, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain.
- Department of Electronics, Universitat de Barcelona, Barcelona 08028, Spain.
| |
Collapse
|
56
|
Heussner A, Paget T. Evaluation of renal in vitro models used in ochratoxin research. WORLD MYCOTOXIN J 2016. [DOI: 10.3920/wmj2015.1975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ochratoxin A (OTA) induces renal carcinomas in rodents with a specific localisation in the S3 segment of proximal tubules and distinct early severe tissue alterations, which have been observed also in other species. Pronounced species- and sex-specific differences in toxicity occur and similar effects cannot be excluded in humans, however precise mechanism(s) remain elusive until today. In such cases, the use of in vitro models for mechanistic investigations can be very useful; in particular if a non-genotoxic mechanism of cancer formation is assumed which include cytotoxic effects. However, potential genotoxic mechanisms can also be investigated in vitro. A crucial issue of in vitro research is the choice of the appropriate cell model. Apparently, the cellular target of OTA is the renal proximal tubular cell; therefore cells from this tissue area are the most reasonable model. Furthermore, cells from affected species should be used and can be compared to cells of human origin. Another important parameter is whether to use primary cultures or to choose a cell line from the huge variety of cell lines available. In any case, important characteristics and quality controls need to be verified beforehand. Therefore, this review discusses the renal in vitro models that have been used for the investigation of renal ochratoxin toxicity. In particular, we discuss the choice of the models and the essential parameters making them suitable models for ochratoxin research together with exemplary results from this research. Furthermore, new promising models such as hTERT-immortalised cells and 3D-cultures are briefly discussed.
Collapse
Affiliation(s)
- A.H. Heussner
- Human and Environmental Toxicology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
- Pharmacy Health and Well-being, University of Sunderland, Sciences Complex, Wharncliffe Street, Sunderland SR1 3SD, United Kingdom
| | - T. Paget
- Pharmacy Health and Well-being, University of Sunderland, Sciences Complex, Wharncliffe Street, Sunderland SR1 3SD, United Kingdom
| |
Collapse
|
57
|
Kalashnikova I, Albekairi N, Ali S, Al Enazy S, Rytting E. Cell Culture Models for Drug Transport Studies. Drug Deliv 2016. [DOI: 10.1002/9781118833322.ch7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
58
|
Affiliation(s)
- Falguni Pati
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory KTH – Royal Institute of Technology Stockholm Schweden
| | - Jesper Gantelius
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory KTH – Royal Institute of Technology Stockholm Schweden
| | - Helene Andersson Svahn
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory KTH – Royal Institute of Technology Stockholm Schweden
| |
Collapse
|
59
|
Pati F, Gantelius J, Svahn HA. 3D Bioprinting of Tissue/Organ Models. Angew Chem Int Ed Engl 2016; 55:4650-65. [PMID: 26895542 DOI: 10.1002/anie.201505062] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Indexed: 12/17/2022]
Abstract
In vitro tissue/organ models are useful platforms that can facilitate systematic, repetitive, and quantitative investigations of drugs/chemicals. The primary objective when developing tissue/organ models is to reproduce physiologically relevant functions that typically require complex culture systems. Bioprinting offers exciting prospects for constructing 3D tissue/organ models, as it enables the reproducible, automated production of complex living tissues. Bioprinted tissues/organs may prove useful for screening novel compounds or predicting toxicity, as the spatial and chemical complexity inherent to native tissues/organs can be recreated. In this Review, we highlight the importance of developing 3D in vitro tissue/organ models by 3D bioprinting techniques, characterization of these models for evaluating their resemblance to native tissue, and their application in the prioritization of lead candidates, toxicity testing, and as disease/tumor models.
Collapse
Affiliation(s)
- Falguni Pati
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Jesper Gantelius
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Helene Andersson Svahn
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
60
|
Wilmer MJ, Ng CP, Lanz HL, Vulto P, Suter-Dick L, Masereeuw R. Kidney-on-a-Chip Technology for Drug-Induced Nephrotoxicity Screening. Trends Biotechnol 2015; 34:156-170. [PMID: 26708346 DOI: 10.1016/j.tibtech.2015.11.001] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/02/2015] [Accepted: 11/04/2015] [Indexed: 02/06/2023]
Abstract
Improved model systems to predict drug efficacy, interactions, and drug-induced kidney injury (DIKI) are crucially needed in drug development. Organ-on-a-chip technology is a suitable in vitro system because it reproduces the 3D microenvironment. A kidney-on-a-chip can mimic the structural, mechanical, transport, absorptive, and physiological properties of the human kidney. In this review we address the application of state-of-the-art microfluidic culturing techniques, with a focus on culturing kidney proximal tubules, that are promising for the detection of biomarkers that predict drug interactions and DIKI. We also discuss high-throughput screening and the challenges for in vitro to in vivo extrapolation (IVIVE) that will need to be overcome for successful implementation.
Collapse
Affiliation(s)
- Martijn J Wilmer
- Department of Pharmacology and Toxicology, Radboudumc, PO Box 9101, Nijmegen, HB 6500 The Netherlands.
| | - Chee Ping Ng
- MIMETAS BV, JH Oortweg 19, Leiden, CH, 2333 The Netherlands
| | | | - Paul Vulto
- MIMETAS BV, JH Oortweg 19, Leiden, CH, 2333 The Netherlands
| | - Laura Suter-Dick
- University of Applied Sciences Northwestern Switzerland, School of Life Sciences, Gründenstrasse 40, 4132 Muttenz, Switzerland
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, CG 3584 The Netherlands
| |
Collapse
|
61
|
Stadler M, Walter S, Walzl A, Kramer N, Unger C, Scherzer M, Unterleuthner D, Hengstschläger M, Krupitza G, Dolznig H. Increased complexity in carcinomas: Analyzing and modeling the interaction of human cancer cells with their microenvironment. Semin Cancer Biol 2015; 35:107-24. [DOI: 10.1016/j.semcancer.2015.08.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/19/2015] [Accepted: 08/21/2015] [Indexed: 02/08/2023]
|
62
|
Kumar A, Starly B. Large scale industrialized cell expansion: producing the critical raw material for biofabrication processes. Biofabrication 2015; 7:044103. [DOI: 10.1088/1758-5090/7/4/044103] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
63
|
Shen C, Zhang G, Wang Q, Meng Q. Fabrication of Collagen Gel Hollow Fibers by Covalent Cross-Linking for Construction of Bioengineering Renal Tubules. ACS APPLIED MATERIALS & INTERFACES 2015; 7:19789-19797. [PMID: 26280545 DOI: 10.1021/acsami.5b05809] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Collagen, the most used natural biomacromolecule, has been extensively utilized to make scaffolds for cell cultures in tissue engineering, but has never been fabricated into the configuration of a hollow fiber (HF) for cell culture due to its poor mechanical properties. In this study, renal tubular cell-laden collagen hollow fiber (Col HF) was fabricated by dissolving sacrificial Ca-alginate cores from collagen shells strengthened by carbodiimide cross-linking. The inner/outer diameters of the Col HF were precisely controlled by the flow rates of core alginate/shell collagen solution in the microfluidic device. As found, the renal tubular cells self-assembled into renal tubules with diameters of 50-200 μm post to the culture in Col HF for 10 days. According to the 3D reconstructed confocal images or HE staining, the renal cells appeared as a tight tubular monolayer on the Col HF inner surface, sustaining more 3D cell morphology than the cell layer on the 2D flat collagen gel surface. Moreover, compared with the cultures in either a Transwell or polymer HF membrane, the renal tubules in Col HF exhibited at least 1-fold higher activity on brush border enzymes of alkaline phosphatase and γ-glutamyltransferase, consistent with their gene expressions. The enhancement occurred similarly on multidrug resistance protein 2 and glucose uptake. Such bioengineered renal tubules in Col HF will present great potential as alternatives to synthetic HF in both clinical use and pharmaceutical investigation.
Collapse
Affiliation(s)
- Chong Shen
- Department of Chemical and Biological Engineering, Zhejiang University , Hangzhou 310027, China
| | - Guoliang Zhang
- College of Chemical Engineering and Materials Science, Zhejiang University of Technology , Hangzhou 310023, China
| | - Qichen Wang
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology , Hoboken, New Jersey 07030, United States
| | - Qin Meng
- Department of Chemical and Biological Engineering, Zhejiang University , Hangzhou 310027, China
| |
Collapse
|
64
|
Batchelder CA, Martinez ML, Duru N, Meyers FJ, Tarantal AF. Three Dimensional Culture of Human Renal Cell Carcinoma Organoids. PLoS One 2015; 10:e0136758. [PMID: 26317980 PMCID: PMC4552551 DOI: 10.1371/journal.pone.0136758] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 08/07/2015] [Indexed: 12/22/2022] Open
Abstract
Renal cell carcinomas arise from the nephron but are heterogeneous in disease biology, clinical behavior, prognosis, and response to systemic therapy. Development of patient-specific in vitro models that efficiently and faithfully reproduce the in vivo phenotype may provide a means to develop personalized therapies for this diverse carcinoma. Studies to maintain and model tumor phenotypes in vitro were conducted with emerging three-dimensional culture techniques and natural scaffolding materials. Human renal cell carcinomas were individually characterized by histology, immunohistochemistry, and quantitative PCR to establish the characteristics of each tumor. Isolated cells were cultured on renal extracellular matrix and compared to a novel polysaccharide scaffold to assess cell-scaffold interactions, development of organoids, and maintenance of gene expression signatures over time in culture. Renal cell carcinomas cultured on renal extracellular matrix repopulated tubules or vessel lumens in renal pyramids and medullary rays, but cells were not observed in glomeruli or outer cortical regions of the scaffold. In the polysaccharide scaffold, renal cell carcinomas formed aggregates that were loosely attached to the scaffold or free-floating within the matrix. Molecular analysis of cell-scaffold constructs including immunohistochemistry and quantitative PCR demonstrated that individual tumor phenotypes could be sustained for up to 21 days in culture on both scaffolds, and in comparison to outcomes in two-dimensional monolayer cultures. The use of three-dimensional scaffolds to engineer a personalized in vitro renal cell carcinoma model provides opportunities to advance understanding of this disease.
Collapse
Affiliation(s)
- Cynthia A. Batchelder
- California National Primate Research Center, University of California Davis, Davis, CA, United States of America
| | - Michele L. Martinez
- California National Primate Research Center, University of California Davis, Davis, CA, United States of America
| | - Nadire Duru
- California National Primate Research Center, University of California Davis, Davis, CA, United States of America
| | - Frederick J. Meyers
- Department of Internal Medicine, School of Medicine, University of California Davis, Davis, CA, United States of America
- Comprehensive Cancer Center, University of California Davis, Davis, CA, United States of America
| | - Alice F. Tarantal
- California National Primate Research Center, University of California Davis, Davis, CA, United States of America
- Comprehensive Cancer Center, University of California Davis, Davis, CA, United States of America
- Department of Pediatrics, School of Medicine, University of California Davis, Davis, CA, United States of America
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California Davis, Davis, CA, United States of America
- * E-mail:
| |
Collapse
|
65
|
Adler M, Ramm S, Hafner M, Muhlich JL, Gottwald EM, Weber E, Jaklic A, Ajay AK, Svoboda D, Auerbach S, Kelly EJ, Himmelfarb J, Vaidya VS. A Quantitative Approach to Screen for Nephrotoxic Compounds In Vitro. J Am Soc Nephrol 2015; 27:1015-28. [PMID: 26260164 DOI: 10.1681/asn.2015010060] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 06/16/2015] [Indexed: 12/16/2022] Open
Abstract
Nephrotoxicity due to drugs and environmental chemicals accounts for significant patient mortality and morbidity, but there is no high throughput in vitro method for predictive nephrotoxicity assessment. We show that primary human proximal tubular epithelial cells (HPTECs) possess characteristics of differentiated epithelial cells rendering them desirable to use in such in vitro systems. To identify a reliable biomarker of nephrotoxicity, we conducted multiplexed gene expression profiling of HPTECs after exposure to six different concentrations of nine human nephrotoxicants. Only overexpression of the gene encoding heme oxygenase-1 (HO-1) significantly correlated with increasing dose for six of the compounds, and significant HO-1 protein deregulation was confirmed with each of the nine nephrotoxicants. Translatability of HO-1 increase across species and platforms was demonstrated by computationally mining two large rat toxicogenomic databases for kidney tubular toxicity and by observing a significant increase in HO-1 after toxicity using an ex vivo three-dimensional microphysiologic system (kidney-on-a-chip). The predictive potential of HO-1 was tested using an additional panel of 39 mechanistically distinct nephrotoxic compounds. Although HO-1 performed better (area under the curve receiver-operator characteristic curve [AUC-ROC]=0.89) than traditional endpoints of cell viability (AUC-ROC for ATP=0.78; AUC-ROC for cell count=0.88), the combination of HO-1 and cell count further improved the predictive ability (AUC-ROC=0.92). We also developed and optimized a homogenous time-resolved fluorescence assay to allow high throughput quantitative screening of nephrotoxic compounds using HO-1 as a sensitive biomarker. This cell-based approach may facilitate rapid assessment of potential nephrotoxic therapeutics and environmental chemicals.
Collapse
Affiliation(s)
- Melanie Adler
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts; Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Susanne Ramm
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Marc Hafner
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Jeremy L Muhlich
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Esther Maria Gottwald
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - Elijah Weber
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Alenka Jaklic
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Amrendra Kumar Ajay
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Scott Auerbach
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Jonathan Himmelfarb
- Kidney Research Institute, Department of Medicine, University of Washington, Seattle, Washington; and
| | - Vishal S Vaidya
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts; Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
66
|
Abstract
The development of safe, effective and patient-acceptable drug products is an expensive and lengthy process and the risk of failure at different stages of the development life-cycle is high. Improved biopharmaceutical tools which are robust, easy to use and accurately predict the in vivo response are urgently required to help address these issues. In this review the advantages and challenges of in vitro 3D versus 2D cell culture models will be discussed in terms of evaluating new drug products at the pre-clinical development stage. Examples of models with a 3D architecture including scaffolds, cell-derived matrices, multicellular spheroids and biochips will be described. The ability to simulate the microenvironment of tumours and vital organs including the liver, kidney, heart and intestine which have major impact on drug absorption, distribution, metabolism and toxicity will be evaluated. Examples of the application of 3D models including a role in formulation development, pharmacokinetic profiling and toxicity testing will be critically assessed. Although utilisation of 3D cell culture models in the field of drug delivery is still in its infancy, the area is attracting high levels of interest and is likely to become a significant in vitro tool to assist in drug product development thus reducing the requirement for unnecessary animal studies.
Collapse
|
67
|
Wick P, Chortarea S, Guenat OT, Roesslein M, Stucki JD, Hirn S, Petri-Fink A, Rothen-Rutishauser B. In vitro-ex vivo model systems for nanosafety assessment. EUROPEAN JOURNAL OF NANOMEDICINE 2015. [DOI: 10.1515/ejnm-2014-0049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractEngineered nanomaterials have unique and novel properties enabling wide-ranging new applications in nearly all fields of research. As these new properties have raised concerns about potential adverse effects for the environment and human health, extensive efforts are underway to define reliable, cost- and time-effective, as well as mechanistic-based testing strategies to replace the current method of animal testing, which is still the most prevalent model used for the risk assessment of chemicals. Current approaches for nanomaterials follow this line. The aim of this review is to explore and qualify the relevance of new in vitro and ex vivo models in (nano)material safety assessment, a crucial prerequisite for translation into applications.
Collapse
|
68
|
Unger C, Kramer N, Walzl A, Scherzer M, Hengstschläger M, Dolznig H. Modeling human carcinomas: physiologically relevant 3D models to improve anti-cancer drug development. Adv Drug Deliv Rev 2014; 79-80:50-67. [PMID: 25453261 DOI: 10.1016/j.addr.2014.10.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 09/02/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022]
Abstract
Anti-cancer drug development is inefficient, mostly due to lack of efficacy in human patients. The high fail rate is partly due to the lack of predictive models or the inadequate use of existing preclinical test systems. However, progress has been made and preclinical models were improved or newly developed, which all account for basic features of solid cancers, three-dimensionality and heterotypic cell interaction. Here we give an overview of available in vivo and in vitro models of cancer, which meet the criteria of being 3D and mirroring human tumor-stroma interactions. We only focus on drug response models without touching models for pharmacokinetic and dynamic, toxicity or delivery aspects.
Collapse
|
69
|
Nam KH, Smith AST, Lone S, Kwon S, Kim DH. Biomimetic 3D Tissue Models for Advanced High-Throughput Drug Screening. ACTA ACUST UNITED AC 2014; 20:201-15. [PMID: 25385716 DOI: 10.1177/2211068214557813] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Indexed: 12/13/2022]
Abstract
Most current drug screening assays used to identify new drug candidates are 2D cell-based systems, even though such in vitro assays do not adequately re-create the in vivo complexity of 3D tissues. Inadequate representation of the human tissue environment during a preclinical test can result in inaccurate predictions of compound effects on overall tissue functionality. Screening for compound efficacy by focusing on a single pathway or protein target, coupled with difficulties in maintaining long-term 2D monolayers, can serve to exacerbate these issues when using such simplistic model systems for physiological drug screening applications. Numerous studies have shown that cell responses to drugs in 3D culture are improved from those in 2D, with respect to modeling in vivo tissue functionality, which highlights the advantages of using 3D-based models for preclinical drug screens. In this review, we discuss the development of microengineered 3D tissue models that accurately mimic the physiological properties of native tissue samples and highlight the advantages of using such 3D microtissue models over conventional cell-based assays for future drug screening applications. We also discuss biomimetic 3D environments, based on engineered tissues as potential preclinical models for the development of more predictive drug screening assays for specific disease models.
Collapse
Affiliation(s)
- Ki-Hwan Nam
- Department of Bioengineering, University of Washington, Seattle, WA, USA Department of Electrical and Computer Engineering, Seoul National University, Seoul, Republic of Korea Center for Analytical Instrumentation Development, The Korea Basic Science Institute, Deajeon, Republic of Korea
| | - Alec S T Smith
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Saifullah Lone
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, Republic of Korea
| | - Sunghoon Kwon
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, Republic of Korea
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA, USA Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
70
|
Huang JX, Blaskovich MA, Cooper MA. Cell- and biomarker-based assays for predicting nephrotoxicity. Expert Opin Drug Metab Toxicol 2014; 10:1621-35. [DOI: 10.1517/17425255.2014.967681] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
71
|
Astashkina AI, Jones CF, Thiagarajan G, Kurtzeborn K, Ghandehari H, Brooks BD, Grainger DW. Nanoparticle toxicity assessment using an in vitro 3-D kidney organoid culture model. Biomaterials 2014; 35:6323-31. [DOI: 10.1016/j.biomaterials.2014.04.060] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/16/2014] [Indexed: 11/16/2022]
|
72
|
Astashkina A, Grainger DW. Critical analysis of 3-D organoid in vitro cell culture models for high-throughput drug candidate toxicity assessments. Adv Drug Deliv Rev 2014; 69-70:1-18. [PMID: 24613390 DOI: 10.1016/j.addr.2014.02.008] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 02/14/2014] [Accepted: 02/18/2014] [Indexed: 12/18/2022]
Abstract
Drug failure due to toxicity indicators remains among the primary reasons for staggering drug attrition rates during clinical studies and post-marketing surveillance. Broader validation and use of next-generation 3-D improved cell culture models are expected to improve predictive power and effectiveness of drug toxicological predictions. However, after decades of promising research significant gaps remain in our collective ability to extract quality human toxicity information from in vitro data using 3-D cell and tissue models. Issues, challenges and future directions for the field to improve drug assay predictive power and reliability of 3-D models are reviewed.
Collapse
|
73
|
DesRochers TM, Palma E, Kaplan DL. Tissue-engineered kidney disease models. Adv Drug Deliv Rev 2014; 69-70:67-80. [PMID: 24361391 DOI: 10.1016/j.addr.2013.12.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 02/08/2023]
Abstract
Renal disease represents a major health problem that often results in end-stage renal failure necessitating dialysis and eventually transplantation. Historically these diseases have been studied with patient observation and screening, animal models, and two-dimensional cell culture. In this review, we focus on recent advances in tissue engineered kidney disease models that have the capacity to compensate for the limitations of traditional modalities. The cells and materials utilized to develop these models are discussed and tissue engineered models of polycystic kidney disease, drug-induced nephrotoxicity, and the glomerulus are examined in detail. The application of these models has the potential to direct future disease treatments and preclinical drug development.
Collapse
|
74
|
Tiong HY, Huang P, Xiong S, Li Y, Vathsala A, Zink D. Drug-induced nephrotoxicity: clinical impact and preclinical in vitro models. Mol Pharm 2014; 11:1933-48. [PMID: 24502545 DOI: 10.1021/mp400720w] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The kidney is a major target for drug-induced toxicity. Drug-induced nephrotoxicity remains a major problem in the clinical setting, where the use of nephrotoxic drugs is often unavoidable. This leads frequently to acute kidney injury, and current problems are discussed. One strategy to avoid such problems would be the development of drugs with decreased nephrotoxic potential. However, the prediction of nephrotoxicity during preclinical drug development is difficult and nephrotoxicity is typically detected only late. Also, the nephrotoxic potential of newly approved drugs is often underestimated. Regulatory approved or validated in vitro models for the prediction of nephrotoxicity are currently not available. Here, we will review current approaches on the development of such models. This includes a discussion of three-dimensional and microfluidic models and recently developed stem cell based approaches. Most in vitro models have been tested with a limited number of compounds and are of unclear predictivity. However, some studies have tested larger numbers of compounds and the predictivity of the respective in vitro model had been determined. The results showed that high predictivity can be obtained by using primary or stem cell derived human renal cells in combination with appropriate end points.
Collapse
Affiliation(s)
- Ho Yee Tiong
- Yong Loo Lin School of Medicine, National University Health System , 1E Kent Ridge Road, NUHS Tower Block, Singapore 119228, Singapore
| | | | | | | | | | | |
Collapse
|
75
|
Li Y, Kandasamy K, Chuah JKC, Lam YN, Toh WS, Oo ZY, Zink D. Identification of Nephrotoxic Compounds with Embryonic Stem-Cell-Derived Human Renal Proximal Tubular-Like Cells. Mol Pharm 2014; 11:1982-90. [DOI: 10.1021/mp400637s] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yao Li
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Karthikeyan Kandasamy
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Jacqueline Kai Chin Chuah
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Yue Ning Lam
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Wei Seong Toh
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Zay Yar Oo
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Daniele Zink
- Institute of Bioengineering
and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| |
Collapse
|
76
|
Fisel P, Renner O, Nies AT, Schwab M, Schaeffeler E. Solute carrier transporter and drug-related nephrotoxicity: the impact of proximal tubule cell models for preclinical research. Expert Opin Drug Metab Toxicol 2014; 10:395-408. [PMID: 24397389 DOI: 10.1517/17425255.2014.876990] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The final excretion step of several drugs is facilitated by membrane transporters of the Solute carrier (SLC) family expressed in the proximal tubules of the kidney. Membrane transporters contribute substantially to the pharmacokinetic profile of drugs and play important roles in drug-induced nephrotoxicity. Different cell models have been applied as tools for the assessment of nephrotoxic effects caused by drugs. AREAS COVERED This review gives an overview over clinically relevant SLC transporters involved in the renal elimination of drug agents and their specific role in drug-induced nephrotoxicity. Most widely applied cell models are described and their advantages and limitations are outlined. EXPERT OPINION In vitro cell culture models (e.g., continuous and primary renal cell lines, polarized cell monolayers) represent valuable tools for early assessment of the nephrotoxic potential of drugs. Since SLC transporters contribute to drug excretion in a large part, in vitro cell culture models might be very helpful to study transport pathways and/or potential drug-drug interactions at an early stage of the drug development process to predict nephrotoxic effects.
Collapse
Affiliation(s)
- Pascale Fisel
- Margarete Fischer-Bosch-Institute of Clinical Pharmacology , Auerbachstrasse 125, Stuttgart, 70376 , Germany
| | | | | | | | | |
Collapse
|
77
|
Hoppensack A, Kazanecki CC, Colter D, Gosiewska A, Schanz J, Walles H, Schenke-Layland K. A human in vitro model that mimics the renal proximal tubule. Tissue Eng Part C Methods 2014; 20:599-609. [PMID: 24266327 DOI: 10.1089/ten.tec.2013.0446] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Human in vitro-manufactured tissue and organ models can serve as powerful enabling tools for the exploration of fundamental questions regarding cell, matrix, and developmental biology in addition to the study of drug delivery dynamics and kinetics. To date, the development of a human model of the renal proximal tubule (PT) has been hindered by the lack of an appropriate cell source and scaffolds that allow epithelial monolayer formation and maintenance. Using extracellular matrices or matrix proteins, an in vivo-mimicking environment can be created that allows epithelial cells to exhibit their typical phenotype and functionality. Here, we describe an in vitro-engineered PT model. We isolated highly proliferative cells from cadaveric human kidneys (human kidney-derived cells [hKDCs]), which express markers that are associated with renal progenitor cells. Seeded on small intestinal submucosa (SIS), hKDCs formed a confluent monolayer and displayed the typical phenotype of PT epithelial cells. PT markers, including N-cadherin, were detected throughout the hKDC culture on the SIS, whereas markers of later tubule segments were weak (E-cadherin) or not (aquaporin-2) expressed. Basement membrane and microvilli formation demonstrated a strong polarization. We conclude that the combination of hKDCs and SIS is a suitable cell-scaffold composite to mimic the human PT in vitro.
Collapse
Affiliation(s)
- Anke Hoppensack
- 1 Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB) , Stuttgart, Germany
| | | | | | | | | | | | | |
Collapse
|
78
|
Kimlin L, Kassis J, Virador V. 3D in vitro tissue models and their potential for drug screening. Expert Opin Drug Discov 2013; 8:1455-66. [PMID: 24144315 DOI: 10.1517/17460441.2013.852181] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The development of one standard, simplified in vitro three-dimensional tissue model suitable to biological and pathological investigation and drug-discovery may not yet be feasible, but standardized models for individual tissues or organs are a possibility. Tissue bioengineering, while concerned with finding methods of restoring functionality in disease, is developing technology that can be miniaturized for high throughput screening (HTS) of putative drugs. Through collaboration between biologists, physicists and engineers, cell-based assays are expanding into the realm of tissue analysis. Accordingly, three-dimensional (3D) micro-organoid systems will play an increasing role in drug testing and therapeutics over the next decade. Nevertheless, important hurdles remain before these models are fully developed for HTS. AREAS COVERED We highlight advances in the field of tissue bioengineering aimed at enhancing the success of drug candidates through pre-clinical optimization. We discuss models that are most amenable to high throughput screening with emphasis on detection platforms and data modeling. EXPERT OPINION Modeling 3D tissues to mimic in-vivo architecture remains a major challenge. As technology advances to provide novel methods of HTS analysis, so do potential pitfalls associated with such models and methods. We remain hopeful that integration of biofabrication with HTS will significantly reduce attrition rates in drug development.
Collapse
Affiliation(s)
- Lauren Kimlin
- 1114 Riverview Terrace, St. Michaels, MD 21663 , USA
| | | | | |
Collapse
|
79
|
Cho EJ, Holback H, Liu KC, Abouelmagd SA, Park J, Yeo Y. Nanoparticle characterization: state of the art, challenges, and emerging technologies. Mol Pharm 2013; 10:2093-110. [PMID: 23461379 DOI: 10.1021/mp300697h] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanoparticles have received enormous attention as a promising tool to enhance target-specific drug delivery and diagnosis. Various in vitro and in vivo techniques are used to characterize a new system and predict its clinical efficacy. These techniques enable efficient comparison across nanoparticles and facilitate a product optimization process. On the other hand, we recognize their limitations as a prediction tool, due to inadequate applications and overly simplified test conditions. We provide a critical review of in vitro and in vivo techniques currently used for evaluation of nanoparticles and introduce emerging techniques and models that may be used complementarily.
Collapse
Affiliation(s)
- Eun Jung Cho
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | | | | | |
Collapse
|
80
|
Bioengineered 3D human kidney tissue, a platform for the determination of nephrotoxicity. PLoS One 2013; 8:e59219. [PMID: 23516613 PMCID: PMC3597621 DOI: 10.1371/journal.pone.0059219] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 02/12/2013] [Indexed: 12/23/2022] Open
Abstract
The staggering cost of bringing a drug to market coupled with the extremely high failure rate of prospective compounds in early phase clinical trials due to unexpected human toxicity makes it imperative that more relevant human models be developed to better predict drug toxicity. Drug–induced nephrotoxicity remains especially difficult to predict in both pre-clinical and clinical settings and is often undetected until patient hospitalization. Current pre-clinical methods of determining renal toxicity include 2D cell cultures and animal models, both of which are incapable of fully recapitulating the in vivo human response to drugs, contributing to the high failure rate upon clinical trials. We have bioengineered a 3D kidney tissue model using immortalized human renal cortical epithelial cells with kidney functions similar to that found in vivo. These 3D tissues were compared to 2D cells in terms of both acute (3 days) and chronic (2 weeks) toxicity induced by Cisplatin, Gentamicin, and Doxorubicin using both traditional LDH secretion and the pre-clinical biomarkers Kim-1 and NGAL as assessments of toxicity. The 3D tissues were more sensitive to drug-induced toxicity and, unlike the 2D cells, were capable of being used to monitor chronic toxicity due to repeat dosing. The inclusion of this tissue model in drug testing prior to the initiation of phase I clinical trials would allow for better prediction of the nephrotoxic effects of new drugs.
Collapse
|
81
|
Li Y, Oo ZY, Chang SY, Huang P, Eng KG, Zeng JL, Kaestli AJ, Gopalan B, Kandasamy K, Tasnim F, Zink D. An in vitro method for the prediction of renal proximal tubular toxicity in humans. Toxicol Res (Camb) 2013. [DOI: 10.1039/c3tx50042j] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
82
|
Gibbons MC, Foley MA, Cardinal KO. Thinking inside the box: keeping tissue-engineered constructs in vitro for use as preclinical models. TISSUE ENGINEERING PART B-REVIEWS 2012; 19:14-30. [PMID: 22800715 DOI: 10.1089/ten.teb.2012.0305] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tissue engineers have made great strides toward the creation of living tissue replacements for a wide range of tissue types and applications, with eventual patient implantation as the primary goal. However, an alternate use of tissue-engineered constructs exists: as in vitro preclinical models for purposes such as drug screening and device testing. Tissue-engineered preclinical models have numerous potential advantages over existing models, including cultivation in three-dimensional geometries, decreased cost, increased reproducibility, precise control over cultivation conditions, and the incorporation of human cells. Over the past decade, a number of researchers have developed and used tissue-engineered constructs as preclinical models for testing pharmaceuticals, gene therapies, stents, and other technologies, with examples including blood vessels, skeletal muscle, bone, cartilage, skin, cardiac muscle, liver, cornea, reproductive tissues, adipose, small intestine, neural tissue, and kidney. The focus of this article is to review accomplishments toward the creation and use of tissue-engineered preclinical models of each of these different tissue types.
Collapse
Affiliation(s)
- Michael C Gibbons
- Department of Biomedical and General Engineering, Cal Poly San Luis Obispo, San Luis Obispo, California 93407, USA
| | | | | |
Collapse
|